Publications

Detailed Information

Immunomodulatory properties and senescence-associated secretory phenotypes in human mesenchymal stem cell aging : 인간 중간엽 줄기세포의 노화에 따른 면역 조절능 및 분비 양상 연구

DC Field Value Language
dc.contributor.advisor강경선-
dc.contributor.author이진영-
dc.date.accessioned2018-11-12T00:57:28Z-
dc.date.available2018-11-12T00:57:28Z-
dc.date.issued2018-08-
dc.identifier.other000000152147-
dc.identifier.urihttps://hdl.handle.net/10371/143152-
dc.description학위논문 (박사)-- 서울대학교 대학원 : 수의과대학 수의학과, 2018. 8. 강경선.-
dc.description.abstractBecause human mesenchymal stem cells (hMSC) have profound immunomodulatory effects, many attempts have been made to use hMSCs in preclinical and clinical trials. For hMSCs to be used in therapy, a large population of hMSCs must be generated by in vitro expansion. However, the immunomodulatory changes following the in vitro expansion of hMSCs have not been elucidated. In the first and second part of this study, I evaluated the effect of senescence on the immunomodulatory ability of hMSCs. Late-passage hMSCs showed impaired suppressive effect on mitogen-induced mononuclear cell proliferation. Interestingly, late-passage hMSCs had a significantly compromised protective effect against mouse experimental colitis, which was confirmed by gross and histologic examination. Among the anti-inflammatory cytokines, the production of prostaglandin E2 (PGE2) and the expression of its primary enzyme, cyclooxygenase-2 (COX-2), were profoundly increased by pre-stimulation with interferon gamma (IFN-γ) and tumor necrosis factor alpha (TNF-α), and this response was significantly decreased with consecutive passages. I demonstrated that the impaired phosphorylation activity of p38 MAP kinase (p38 MAPK) in late-passage hMSCs led to a compromised immunomodulatory ability through the regulation of COX-2. These results indicate that the immunomodulatory ability of hMSCs gradually declines with consecutive passages via a p38-mediated alteration of COX-2 and PGE2 levels.

Low oxygen environment have been reported to inhibit senescence and maintain undifferentiated states in somatic stem cells. In the second part of study, I evaluated the effects of hypoxia on cellular senescence and the immunomodulatory abilities of hMSCs. Hypoxic-cultured human umbilical cord blood-derived mesenchymal stem cells (hUCB-MSCs) showed enhanced proliferation and had increased immunosuppressive effects on mitogen-induced mononuclear cell proliferation. I found that BMI1, a member of the polycomb repressive complex protein group, showed increased expression in hypoxic-cultured hUCB-MSCs, and the further knock down of BMI1 in hypoxic cells induced decreased proliferative and immunomodulatory abilities in hUCB-MSCs, along with COX-2/PGE2 down-regulation. Furthermore, the expression of phosphorylated p38 MAP kinase increased in response to the over-expression of BMI1 in normoxic conditions, suggesting that BMI1 regulates the immunomodulatory properties of hUCB-MSCs via p38 MAP kinase-mediated COX-2 expression. More

importantly, I identified BMI1 as a direct repressor of MAP kinase phosphatase-1 (MKP-1)/DUSP1, which suppresses p38 MAP kinase activity. The results demonstrate that hypoxia induced BMI1 plays a key role in the regulation of the immunomodulatory properties of hMSCs.

Defects in the nuclear lamina occur during physiological aging and as a result of premature aging disorders. Aging is also accompanied by an increase in transcription of genes encoding cytokines and chemokines, a phenomenon known as the senescence-associated secretory phenotype (SASP). Progerin and prelamin A trigger premature senescence and loss of function of hMSCs, but little is known about how defects in nuclear lamin A regulate SASP. In the third part of study, I show that both progerin overexpression and ZMPSTE24 depletion induce paracrine senescence, especially through expression of monocyte chemoattractant protein-1 (MCP-1), in hMSCs. Importantly, I identified that GATA binding protein 4 (GATA4) is a mediator regulating MCP-1 expression in response to prelamin A or progerin in hMSCs. Co-immunoprecipitation revealed that GATA4 expression is maintained due to impaired p62-mediated degradation in progerin-expressing hMSCs. Furthermore, depletion of GATA4 abrogated SASP-dependent senescence through suppression of NF-ĸB and MCP-1 in hMSCs with progerin or prelamin A. Thus, my findings indicate that abnormal lamin A proteins trigger paracrine senescence through a GATA4-dependent pathway in hMSCs. This molecular link between defective lamin A and GATA4 can provide insights into physiological aging and pathological aging disorders.

In conclusion, cellular senescence can alter immunomodulatory abilities and paracrine senescence phenotypes in hMSCs. The molecular mechanisms demonstrated in this study can provide insights into physiological aging and pathological aging disorders. These findings also suggest a strategy to enhance the functionality of hMSCs for use in therapeutic applications.
-
dc.description.tableofcontentsCHAPTER I: A p38 MAPK-mediated alteration of COX-2/PGE2 regulates immunomodulatory properties in human mesenchymal stem cell aging 1

1.1 INTRODUCTION 2

1.2 MATERIALS AND METHODS 5

1.2.1 Cell culture 5

1.2.2 Flow cytometric analysis 5

1.2.3 In vitro differentiation assay 6

1.2.4 Isolation and culture of hUCB-derived MNCs 6

1.2.5 Senescence-associated betagalactosidase (SA β-gal) staining 7

1.2.6 Cumulative population doubling level 7

1.2.7 Mixed Lymphocyte Reaction (MLR) 8

1.2.8 Colitis induction 8

1.2.9 Cytokine production 10

1.2.10 Western blot analysis 10

1.2.11 Immunocytochemistry 11

1.2.12 Statistical analysis 11

1.3 RESULTS 12

1.3.1 Characterization of hMSCs 12

1.3.2 Replicatively senescent hMSCs show senescence phenotypes with a compromised immunosuppressive ability 15

1.3.3 Early- passage hMSCs show enhanced protective effects against DSS-induced colitis in mice 18

1.3.4 Alteration of immunomodulatory activities during hMSC aging is dependent on PGE2 production 22

1.3.5 p38 MAP kinase is responsible for the reduced expression of COX-2 26

1.4 DISCUSSION 28



CHAPTER Ⅱ: BMI1 inhibits senescence and enhances the immunomodulatory properties of human mesenchymal stem cells via the direct suppression of MKP-1/DUSP 1 32

2.1 INTRODUCTION 33

2.2 MATERIALS AND METHODS 36

2.2.1 Cell culture 36

2.2.2 Characterization of hUCB-MSCs 36

2.2.3 In vitro differentiation assay 37

2.2.4 Cumulative population doubling level (CPDL) 37

2.2.5 Senescence-associated beta-galactosidase (SA-ß-gal) staining 38

2.2.6 MTT assay 38

2.2.7 Western blot analysis 39

2.2.8 Isolation and culture of hUCB-derived MNCs 39

2.2.9 MNC proliferation assay 40

2.2.10 Retroviral transduction 40

2.2.11 Chromatin immunoprecipitation 41

2.2.12 Statistical analysis 41

2.3 RESULTS 43

2.3.1 Hypoxic culturing decreases cellular senescence in hUCB-MSCs with increased BMI1 expression 43

2.3.2 Hypoxia-induced BMI1 regulates cellular senescence in hUCB-MSCs 48

2.3.3 BMI1 regulates the immunosuppressive properties of hUCB-MSCs in hypoxia 54

2.3.4 BMI1 upregulation enhances the immunomodulatory properties of hUCB-MSCs 58

2.3.5 BMI1 inhibits MKP-1/DUSP1 expression in hypoxia-cultured hUCB-MSCs 62

2.3.6 BMI1 directly inhibits MKP-1/DUSP1 expression by binding to the promoter of DUSP1 65

2.4 DISCUSSION 67



CHAPER Ⅲ: GATA4-dependent regulation of the secretory phenotype via MCP-1 underlies lamin A-mediated human mesenchymal stem cell aging 71

3.1 INTRODUCTION 72

3.2 MATERIALS AND METHODS 75

3.2.1 Cell culture 75

3.2.2 Isolation and culture of hUCB-derived MNCs 75

3.2.3 Senescence-associated beta-galactosidase (SA-β-gal) staining 75

3.2.4 MTT assay 76

3.2.5 Western blot analysis 76

3.2.6 Conditioned Medium (CM) 77

3.2.7 MNC migration assay 77

3.2.8 Viral transduction 77

3.2.9 Quantitative RT-PCR 78

3.2.10 Immunocytochemistry 79

3.2.11 Co-immunoprecipitation 79

3.2.12 Statistical analysis 80

3.3 RESULTS 82

3.3.1 Progerin induces senescence phenotypes in hUCB-MSCs 82

3.3.2 Progerin induces SASP via MCP-1 expression 86

3.3.3 GATA4 regulates SASP caused by progerin in hMSCs 89

3.3.4 Depletion of GATA4 inhibits senescence in progerin-over-expressing hMSCs 93

3.3.5 GATA4 down-regulation decreases senescence induced by prelamin A 97

3.4 DISCUSSION 105



GENERAL CONCLUSION 108

REFERENCES 111

ABSTRACT IN KOREAN 140


-
dc.language.isoen-
dc.publisher서울대학교 대학원-
dc.subject.ddc636.089-
dc.titleImmunomodulatory properties and senescence-associated secretory phenotypes in human mesenchymal stem cell aging-
dc.title.alternative인간 중간엽 줄기세포의 노화에 따른 면역 조절능 및 분비 양상 연구-
dc.typeThesis-
dc.description.degreeDoctor-
dc.contributor.affiliation수의과대학 수의학과-
dc.date.awarded2018-08-
Appears in Collections:
Files in This Item:

Altmetrics

Item View & Download Count

  • mendeley

Items in S-Space are protected by copyright, with all rights reserved, unless otherwise indicated.

Share